You are on page 1of 16
sariv2021 CCancer-Promoting fects of Microbial Dysbiosis HHS Public Access Lk Author manuscript Peer-reviewed and asoepted for publication ETE EMT Curr Oncol Rep. Author manuscript; available in PMC 2015 Oct 4 PMCID: PMc4180221 Published in final edited form as: NIHMSID: NIHMS621515 Pup: 25123079 Cancer-Promoting Effects of Microbial Dysbiosis Amy M. Shefin,’ Alyssa K. Whitney,” and Tiffany L. Weit!* ‘Department of Food Science and Human Nutrition, Colorado State University, Fort Collins, CO 80523 2Department of Biology, Colorado State University, Fort Collins, CO 80523, “author to whom correspondence should be addressed: Tiffany, Welr@colostate,edu Copyright notice Abstract Humans depend upon our commensal bacteria for nutritive, immune-modulating and metabolic contributions to maintenance of health. However, this commensal community exists in careful balance that, if disrupted, enters dysbiosis; which has been shown to contribute to the etiology of colon, gastric, esophageal, pancreatic, laryngeal, breast and gallbladder carcinomas. This etiology is closely tied to host inflammation, which causes and is aggravated by microbial dysbiosis while increasing vulnerability to pathogens. Advances in sequencing technology have increased our ability to catalog microbial species associated with various cancer types across the body. However, defining microbial biomarkers as cancer predictors presents multiple challenges and existing studies identifying cancer- associated bacteria have reported inconsistent outcomes. Combining metabolites and microbiome analyses can help elucidate interactions between gut microbiota, metabolism and the host. Ultimately, understanding how gut dysbiosis impacts host response and inflammation will be critical to creating an accurate picture of the role of the microbiome in canecr. Keywords: microbiome, cancer, microbial metabolites, dysbiosis, inflammation, genotoxins Introduction The relationship between specific pathogenic bacteria and human carcinogenesis has been the subject of extensive investigation, Historically, most of this research has focused on individual pathogens, such as Helicobacter pylori, and their potential to initiate and perpetuate disease. Previous research focus ‘was on the disease process rather than beneficial gut-microbe interactions. More recently, extensive research supports commensal bacteria playing a role in protection of host health via nutritive, immune- modulating and metabolic processes [1, 2]. In addition, the more holistic approach of characterizing entire communities of gut bacteria and their interactions is now possible through use of high- throughput DNA sequencing technology. Characterization of the gut microbiome as a whole has, furthered our understanding of intestinal microbial ecology to include community-level functions and changes. In healthy individuals, the gut microbiome functions as a symbiont that can offer protection from invading pathogens and prevent tumorigenesis [3]. However, this commensal community exists in careful balance that, if disrupted, enters dysbiosis and contributes to host disease processes, including ‘cancer [4-7]. While recent findings still support individual microorganisms influencing carcinogenesis, hitpsiwowcnebl nim, nin govipmelartles/PMC#180221/ we sariv2021 Ccance-Promating Etfects of Mlabal Dysbisis greater emphasis is on microbial dysbiosis and its larger role in cancer initiation and progression. The focus of this review is on gut microbial community dynamics that shift state from symbiosis to dysbiosis and the subsequent host immune and pathogen response, which drastically alters initiation and progression of multiple types of cancer. Proposed mechanisms for microbiome involvement in colorectal cancer Multiple studies report different gut microbiome composition in individuals diagnosed with colorectal cancer (CRC) versus healthy individuals [8-11]. In fact, gut microbiota can play a role in either promotion or prevention of CRC, often through modulation of the inflammatory process due to close contact with host colonic mucosa [5]. For example, chemically induced injury and proliferation induced by azoxymethane (AOM) and dextran sulfate sodium (DSS) was enhanced in germ free mice, which lack protective commensals. In addition, tumor development in the germ-free mice resulted in significantly more and larger tumors compared to specific pathogen free mice [12]. Balance of the gut microbial community, or eubiosis, can be disrupted by an inflammatory environment in the host. For example, host inflammation may influence microbiota composition through generation of specific metabolites such as nitric oxide synthase (NOS2), Nitrate provides a unique energy source for facultative anaerobic bacteria allowing them to outcompete bacteria that cannot utilize nitrates [13], disrupting balance of the gut microbiome and resulting in dysbiosis. Pro-inflammatory responses can also compromise barrier and immune function to allow bacterial translocation through intestinal tight Junctions and intensify the inflammatory response [14] How inflammation interacts with the gut microbiome to influence CRC has been recently synthesized in several hypotheses that summarize our understanding of the interactions to date (igure 1). The “alpha-bug’ hypothesis suggests that a keystone pathogen species, such as Enterotoxigenic B. fragilis (ETBP), remodels the colonic microbiota to promote CRC, possibly via IL-17 and T}17 cell-mediated inflammation. 1 is process could also be initiated by microbial-independent host-mediated inflammation and may be blocked by beneficial commensal microbiota [15]. Similarly, the bacterial driver-passenger model suggests that ‘driver’ bacteria, such as ETBF, cause or aggravate inflammation and produce genotoxins that lead to cell proliferation and mutations. Subsequently, an adenoma forms and is colonized by ‘passenger’ bacteria such as Fusobacterium spp. that encourage tumor progression [16]. Following tumor formation, the intestinal barrier is damaged by the continual inflammation and allows bacteria access to tumor tissue. These bacteria and their metabolites stimulate additional inflammatory signals, including IL-17 eytokines, promoting cancer progression [17]. Inflammatory signals may also stimulate macrophages, via induction to an M1 phenotype, to produce chromosome- breaking factors through a bystander effect, damaging DNA and inducing chromosomal instability in neighboring cells (3). Likely CRC initiation and progression is engendered by aspects of cach of these models. hitpsiwowcnebl nim, nin govipmelartles/PMC#180221/ 26 sariv2021 CCancer-Promoting Elects of Mlerblal Dysbosis, fefetefefatetel community host —_Keystone-pathogenbacleral baclenal-arver 5° JY homeostasis inflammation active yg rectuting

2 show altered apoptosis and differentiation with Helicobacter {felis infection. Gastroenterology. 1998;114(4):675-689, [PubMed] [Google Scholar} 46, Moen EL, Wen S, Anwar T, Cross-Knorr S, Brilliant K, Birnbaum F, Rahaman S, Sedivy JM, Moss SF, Chatterjee D, Regulation of RKIP function by Helicobacter pylori in gastric cancer, PloS one. 2012;7(5):e37819. [PMC free article} [2ubMed] [Google Scholar] 47. Yang L, Chaudhary N, Baghdadi J, Pei Z. Microbiome in Reflux Disorders and Esophageal Adenocarcinoma, The Cancer Journal, 2014;20(3):207-210, [PMC five article] [PubMed] [Google Scholar 48, Anderson LA, Murphy SJ, Johnston BT, Watson R, Ferguson H, Bamford KB, Ghazy A, McCarron P, McGuigan J, Reynolds JV. Relationship between Helicobacter pylori infection and gastric atrophy and the stages of the oesophageal inflammation, metaplasia, adenocarcinoma sequence: results from the FINBAR case-control study. Gut, 2008;57(6):734~739. [PubMed] [Google Scholar] 49, Yang L, Lu X, Nossa CW, Francois F, Peck RM, Pei Z. Inflammation and intestinal metaplasia of the distal esophagus are associated with alterations in the microbiome. Gastroenterology. 2009;137(2):588-597. [PMC free article] [PubMed] [Google Scholar] hitpsuiwowcnebl nim, govipmelatles/PMC#180221/ 1286 sariv2021 Ccance-Promating Eifects of Mlrabal Dysbisis 50, Narikiyo M, Tanabe C, Yamada Y, Igaki H, Tachimori Y, Kato H, Muto M, Montesano R, Sakamoto H, Nakajima Y, Frequent and preferential infection of Treponema denticola, Streptococcus mitis, and Streptococcus anginosus in esophageal cancers, Cancer science. 2004;95(7):569-574. [PubMed] [Google Scholar] Sis, Farrell 1J, Zhang L, Zhou H, Chia D, Elashoff D, Akin D, Paster BJ, Joshipura K, Wong DT. Variations of oral microbiota are associated with pancreatic diseases including pancreatic cancer. Gut 2012;61(4):582-S88. This study indicates that changes of the oral microbiota may be useful biomarkers in the detection of pancreatic cancer. It provides an illustration of microbial changes between cancer patients and healthy controls, indicating species that are relevant to the development of pancreatic cancer. [PMC five article] [PubMed] [Google Scholar} 52s, Gong HL, Shi ¥, Zhou L, Wa CP, Cao PY, Tao L, Xu C, Hou DS, Wang YZ. The composition of microbiome in larynx and the throat biodiversity between laryngeal squamous cell carcinoma patients and control population. PloS one. 2013;8(6):c66476. This study provides a comprehensive depiction of ‘microbial changes between cancer patients and healthy controls, implicating H. pylori and a number of other species in the development of laryngeal carcinoma, [PMC five article] [PubMed] [Google Scholar 53. Sharma V, Chauhan VS, Nath G, Kumar A, Shukla VK. Role of bile bacteria in gallbladder carcinoma, Hepato-gastroenterology. 2007;54(78):1622. [PubMed] [Google Scholar] 54, Shukla V, Tiwari S, Roy S. Biliary bile acids in cholelithiasis and carcinoma of the gall bladder. European journal of cancer prevention. 1993;2(2):155-160. [PubMed] [Google Scholar] 55, Walboomers JM, Jacobs MV, Manos MM, Bosch FX, Kummer JA, Shah KY, Snijders PJ, Peto J, Meijer CJ, Munoz N. Human papillomavirus is a necessary cause of invasive cervical cancer worldwide. The Journal of pathology. 1999;189(1):12-19. [PubMed] (Google Scholar] 56. Mitrani-Rosenbaum S, Tsvieli R, TurKaspa R. Oestrogen stimulates differential transcription of ‘human papillomavirus type 16 in SiHla cervical carcinoma cells. Journal of general virology. 1989;70(8):2227-2232. [PubMed] [Google Scholar] 57. Riley RR, Duensing 8, Brake T, Miinger K, Lambert PF, Arbeit JM. Dissection of human papillomavirus E6 and E7 function in transgenic mouse models of cervical carcinogenesis. Cancer research. 2003;63(16):4862-4871. [PubMed] [Google Scholar] 58, Chung SH, Wiedmeyer K, Shai A, Korach KS, Lambert PF, Requirement for estrogen receptor a in a mouse model for human papillomavirus-associated cervical cancer. Cancer research. 2008;68(23):9928-9934. [PMC free article] [PubMed] [Google Scholar} 59, Elson DA, Riley RR, Lacey A, Thordarson G, Talamantes FJ, Arbeit IM, Sensitivity of the cervical ‘transformation zone to estrogen-induced squamous carcinogenesis. Cancer research. 2000;60(5):1267— 1275. [PubMed] [Google Scholar 60, Lombardi P, Goldin B, Boutin E, Gorbach SL. Metabolism of androgens and estrogens by human fecal microorganisms, Journal of steroid biochemistry. 1978;9(8):795-801. [PubMed] [Google Scholar] 61, D'Souza G, Kreimer AR, Viscidi R, Pawlita M, Fakhry C, Koch WM, Westra WH, Gillison ML, Case-control study of human papillomavirus and oropharyngeal cancer. New England Journal of Medicine. 2007;356(19):1944—1956. [PubMed] [Google Scholar] 62. Coussens LM, Werb Z. Inflammation and cancer. Nature. 2002;420(6917):860-867. (EMC free article] [PubMed] {Google Scholar] hitpsiwowcnebl nim, nin govipmelartles/PMC#180221/ 1916

You might also like